Abstract
Cytokines are major drivers of autoimmunity, and biologic agents targeting cytokines have revolutionized the treatment of immune-mediated diseases. Despite the effectiveness of these drugs, they do not induce complete remission in all patients, prompting the development of alternative strategies—including targeting of intracellular signal transduction pathways downstream of cytokines. Many cytokines that bind type I and type II cytokine receptors are critical regulators of immune-mediated diseases and employ the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway to exert their effect. Pharmacological inhibition of JAKs block the actions of type I/II cytokines, and within the past 3 years therapeutic JAK inhibitors, or Jakinibs, have become available to rheumatologists. Jakinibs have proven effective for the treatment of rheumatoid arthritis and other inflammatory diseases. Adverse effects of these agents are largely related to their mode of action and include infections and hyperlipidemia. Jakinibs are currently being investigated for a number of new indications, and second-generation selective Jakinibs are being developed and tested. Targeting STATs could be a future avenue for the treatment of rheumatic diseases, although substantial challenges remain. Nonetheless, the ability to therapeutically target intracellular signalling pathways has already created a new paradigm for the treatment of rheumatologic disease.
Introduction
A fundamental insight gained over more than three decades is that diverse cytokines are pivotal drivers of rheumatoid arthritis (RA) and other autoimmune diseases1–3. Cytokine-targeting therapies developed on the basis of these discoveries have dramatically changed the treatment options for patients with RA, psoriasis, inflammatory bowel disease (IBD) and other disorders4–6 . Clearly, however, not all patients respond adequately to these therapies, complete remission is not as common as one would hope, and ‘cure’ remains an elusive goal. These challenges suggest that alternatives to targeting cytokines extracellularly should be considered. Indeed, the prospect of targeting intracellular pathways activated by cytokines is now a reality.
In considering the cytokines that drive autoimmunity, it is important to understand that the term ‘cytokine’ encompasses structurally distinct factors that bind cellular receptors belonging to at least seven families, which signal through very different pathways (Figure 1). In this Review, we focus on cytokines that drive autoimmune disease and that employ one particular pathway, the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway7, 8. Despite their importance in host defense and immune-mediated disease, cytokines such as TNF, IL-1 and IL-17 do not signal via JAKs and STATs. However, JAK–STAT-dependent cytokines can induce these cytokines and in this way also affect immune-mediated disease. In the first part of the Review, we will discuss the roles of JAK-dependent cytokines in normal and aberrant immune responses, and then briefly review JAK-STAT signalling. The second portion of the Review focuses on the emergence of therapies targeting the JAK-STAT signalling pathway.
Figure 1.
Cytokines are grouped into superfamilies based on shared structural elements of the receptors they bind. The major cytokine families are: the type I/II cytokines, the IL-1 family, the TNF family, the IL-17 cytokines, the RTK/SCF cytokines, the TGF-β family cytokines, and chemokines. The type I/II cytokine families signal through JAKs and STATs. When a cytokine binds to its cognate receptor, the receptor becomes activated. JAKs autophosphorylate and transphosphorylate, causing STATs to be recruited to the activated receptor where they, in turn, are phosphorylated. The STATs then dimerize and translocate to the nucleus where they initiate transcription. FDA-approved medications blocking the different classes of cytokine receptor are denoted in red.
Abbreviations: JAK, Janus kinase; RTF, receptor tyrosine kinase; SCF, stem cell factor; STAT, TGF-β, transforming growth factor β.
Roles of Type I/II Cytokines in Health and Disease
Cytokines that bind type I and type II receptors include interleukins, interferons, interferon-like cytokines, colony-stimulating factors, hormones, and growth factors (Table 1). Some of these cytokines share receptor subunits and can be grouped on this basis.
Type I cytokines in autoimmunity
The common γ-chain (γc, also known as IL-2 receptor γ subunit) cytokines include IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The fundamental role of these cytokines in lymphocyte development is illustrated by primary immunodeficiency that arises from inactivating mutations of γc (encoded by IL2RG), termed X-linked severe combined immunodeficiency (X-SCID). Many γc cytokines also have been implicated in diverse autoimmune diseases. IL-2 is the prototypic T-cell growth factor and has both proinflammatory and anti-inflammatory effects. This cytokine is important for effector T cells, upregulating production of other cytokines and augmenting the cytolytic activity of CD8 T cells and natural killer (NK) cells. Blockade of the IL-2 receptor with daclizumab is approved for the treatment of transplant rejection and seems to be efficacious in multiple sclerosis (MS)9, 10. IL-4 is found at elevated levels in the synovial fluid of patients with early RA but is absent in late disease11; generally, though, IL-4 is thought to be more important in driving allergic diseases. Both IL-7 and IL-15 are important for lymphocyte homeostasis and T-cell memory11, 12 IL-15 also regulates NK cells, whereas IL-7 is important for the homeostasis of other innate lymphoid cells13. Thymic stromal lymphopoietin (TSLP) shares the IL-7 receptor alpha chain as a receptor component; it has a role in T cell development and allergy12. IL-9 is involved in atopy and IBD14. IL-21 promotes differentiation of follicular helper T (TFH) cells, which act on B cells in germinal centres to promote antibody class-switching15. Inactivating mutations of IL21 and IL21R result in immunodeficiency16. Because of their role in class switching, TFH cells are thought to be important for diverse autoantibody-linked autoimmune diseases including RA and systemic lupus erythematosus (SLE). Levels of IL-21 are increased in the synovium and serum of patients with RA17. Targeting IL-21 has been investigated in phase 1 trials of RA and Crohn’s disease although results have not yet been reported18.
The common β-chain cytokines include IL-3, IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Encoded by CSF2, GM-CSF is a proinflammatory growth factor with a key role in the generation, survival, and activation of myeloid cells. It is produced by pathogenic T cells, and a body of data points to its importance in driving autoimmunity19. GM-CSF blockade has been effective for RA in phase 2 clinical trials20.
IL-6 is the prototypic proinflammatory cytokine, which, like a number of related cytokines, signals through the gp130 receptor subunit (encoded by IL6ST). Among its many actions, IL-6 drives the production of TNF and IL-1 and induces the differentiation of T helper (TH)17 cells. These cells are the major producers of IL-17, which acts synergistically with TNF to promote activation of chondrocytes and fibroblast-like synoviocytes, production of metalloproteinases, and joint destruction in RA1. Tocilizumab and many other agents in development target IL-6 (Table I). Clinical trials are ongoing for multiple indications beyond RA, such as vasculitis and myositis (ClinicalTrials.gov identifiers NCT01450137 and NCT02043548, respectively)21. Other gp130 cytokines include IL-11, IL-27, and IL-31. IL-11 is involved in haematopoiesis and bone remodelling. IL-31 is produced by TH2 cells, mast cells and other leukocytes; this cytokine has a role in skin barrier function and myeloid development22. IL-27 is also a gp130 cytokine but primarily limits inflammation, including antagonizing IL-17 production23–25 (see below)
Table I.
Selected Type I/II Cytokines Important for Hematopoiesis, Immune Homeostasis, and Immune-mediated Disease
| TYPE I CYTOKINES | Signaling JAKs | Signaling STATs | Immune function and potential role in Immune mediated Disease | Blocking Agent (FDA-Approved or in late phase Clinical Trials) Clinical Indication |
|---|---|---|---|---|
| Common γc | ||||
| IL-2 | JAK1, JAK3 | 3+5 | Enhances effector and regulatory responses | Daclizumab Transplant, MS |
| IL-4 | JAK1, JAK3 | 6 | Allergies and asthma – Th2-mediated diseases | Dupilumab* Asthma |
| IL-7 | JAK1, JAK3 | 3+5 | T cell development and homeostasis | Not applicable |
| IL-9 | JAK1, JAK3 | 1+3+5 | Atopic disease, IBD | Not applicable |
| IL-13** | JAK1, JAK3, JAK2 (???),TYK2 | 6 | Airway hyperresponsiveness, goblet cell metaplasia and mucus hypersecretion, fibrosis | Lebrikizumab Asthma Dupilumab* Asthma |
| IL-15 | JAK1, JAK3 | 3+5 | Memory T cells, NK cells, induction of cell proliferation | Not applicable |
| IL-21 | JAK1, JAK3 | 1+3+5 | Tfh cells | Not applicable |
| Common βχ | ||||
| GM-CSF | JAK2 | 3+5 | Macrophages, T cells, mast cells, NK cells, endothelial cells and fibroblasts, stem cell stimulation and differentiation | Not applicable |
| IL-3 | JAK2 | 3+5+6 | Differentiation of multipotent hematopoietic stem cells, proliferation of all cells in the myeloid lineage | Not applicable |
| IL-5 | JAK2 | 3+5+6 | Allergies, asthma, eosinophilic disease | Mepolizumab Asthma Benralizumab, reslizumab |
| gp130 | ||||
| IL-6 | JAK1, JAK2, TYK2 | 1+3 | Prototypic proinflammatory cytokine Broadly relevant for many autoimmune disease |
Tocilizumab120 RA, sJIA, pJIA Sarilumab, ALX-0061, Olokizumab, Sirukinumab, Siltuximab, Clazakinumab |
| IL-11 | JAK1, JAK2, TYK2 | 3 | Hematopoiesis, bone remodeling | Not applicable |
| IL-27 | JAK1, JAK2, TYK2 | 1+2+3+4+5 | Regulation of the activity of B- and T-lymphocytes | Not applicable |
| Dimeric | ||||
| IL-12 | JAK2, TYK2 | 4 | Th1-associated disease | Ustekinumab ** Psoriasis, PsA |
| IL-23 | JAK2, TYK2 | 3+4 | Th17-mediated disease | Ustekinumab,** Psoriasis, PsA Tildrakizumab, guselkumab |
| IL-27 | JAK1, JAK2 | 1+3 | Enhance Th1 and inhibit Th17 responses | Not applicable |
| IL-35 | JAK1, JAK2 | 1+4 | Anti-inflammatory responses | Not applicable |
| Hormone-like | ||||
| EPO | JAK2 | 5 | Control of erythropoiesis | Adverse effects of Jakinibs |
| TPO | JAK2 | 1+3+5 | Regulation of the differentiation of megakaryocytes and platelets | Adverse effects of Jakinibs |
| G-CSF | JAK2 | 5 | Bone marrow stimulation to produce stem cells and granulocytes | Adverse effects of Jakinibs |
| Growth hormone | JAK2 | 3+5a | Stimulation of cell division of chondrocytes and IGF-1 | Not applicable |
| Leptin | JAK2 | 3+5a | Coordination of energy homeostasis, increases satiety | Not applicable |
| TYPE II CYTOKINES | Signaling JAKs | Signaling STATs | Immune function and potential role in Immune mediated Disease | Blocking Agent or Agonist (FDA-Approved or in late phase Clinical Trials) Clinical Indication |
| IFN family | ||||
| IFNα/β | JAK1, TYK2 | 1+2+3+4+5 | Enhance immunity against infections and drive autoimmunity | Anifrolumab Medi-546 Systemic Sclerosis SLE |
| IFNγ | JAK1, JAK2 | 1 | Enhance immunity against infections and drive autoimmunity | Not applicable |
| IL-28 | JAK1, TYK2 | 1+2-3-4-5 | Enhance immunity against infections | Not applicable |
| IL-29 | JAK1, TYK2 | 1+2-3-4-5 | Enhance immunity against infections | Not applicable |
| IL-10 family | ||||
| IL-10 | JAK1, JAK2, TYK2 | 1+3+5 | Anti-inflammatory actions | Not applicable |
| IL-19 | JAK1, JAK2, TYK2 | 3 | B cell activation Antibody production |
Not applicable |
| IL-20 | JAK1, JAK2, TYK2 | 3 | Synoviocyte migration Osteoclast formation |
Not applicable |
| IL-22 | JAK1, JAK2, TYK2 | 1+3+5 | Promote barrier immunity Augment IL-17 function |
Not applicable |
Table I provides examples of key Type I/II cytokines, the JAKs and STATs through which they signal, and selected targeted therapies that block their actions, and are either FDA-approved or in late phase clinical trials for rheumatic or other diseases. Cytokines are grouped based on shared structure/subunits.
Of note, IL-13 is grouped with the gamma chain cytokines, but IL-13 does not share the common γ χηαιn; rather it shares the IL-4 receptor alpha subunit (IL4Rα). Dupilumab targets IL4Rα and therefore blocks both IL-4 and IL-13.
IL-12 and IL-23 both are composed of p40 subunit. Ustekinumab, which targets p40, blocks both cytokines.
Abbreviations: RA = rheumatoid arthritis, AS – ankylosing spondylitis, HLH = hemophagocytic lymphohistiocytosis, sJIA = systemic juvenile idiopathic arthritis, pJIA = polyarticular juvenile idiopathic arthritis, MS = multiple sclerosis, AML = acute myelogenous leukemia, MDS = myelodysplastic syndroems, HES = hypereosinophilic syndrome, AD = atopic dermatitis, EE = eosinophilic esophagitis, GPA = granulomatosis with polyangiitis, eGPA = eosinophilic granulomatosis with piolyangiitis, COPD = chronic obstructive pulmonary disease, LVV = large vessel vasculitis, GVHD = Graft-versus-host disease, PsA = psoriatic arthritis, UC = ulcerative colitis, IBD = inflammatory bowel disease, SpA = spondyloarthropathy, Ssc = systemic sclerosis, pSS = primary Sjogren’s syndrome, OA = osteoarthritis, HIV = human immunodeficiency virus, IPF = idiopathic pulmonary fibrosis.
Clinical trials can be found at www.clinicaltrials.gov: NCT01463644, NCT00538434, NCT00527566, NCT02238353, NCT02101333, NCT02034474, NCT01297699, NCT02453256, NCT01209689, NCT02477059, NCT01782235, NCT01649375, NCT01961609, NCT01806597, NCT01640938, NCT01392326, NCT01266811, NCT01663441, NCT01369342, NCT02407236, NCT02407223, NCT01274338, NCT02199327, NCT00002882, NCT00003924, NCT00125814, NCT00167583, NCT00002238, NCT00249860, NCT00344253, NCT00070187, NCT00538811, NCT02415127, NCT00076635, NCT01270490, NCT00004402
Related to the gp130 cytokines is the dimeric cytokine family, which includes IL‐12, IL-23, and IL-35. IL-12 promotes the differentiation of IFN-γ-producing TH1 cells, whereas IL-23 promotes IL-17 production and generation of TH17 cells. IL-23 is important in the pathogenesis of psoriasis, IBD, and spondyloarthritis2. Targeting IL-23 and IL-12 with ustekinumab is effective for psoriasis26 and has shown promise as a treatment for IBD27.
Type II cytokines in autoimmunity
The type II cytokines comprise a large group of >30 signalling molecules including the type I, II, and III interferons (IFN-ασ, IFN-β, IFN- γ, IL-28, and IL-29) and IL-10-related cytokines (IL-10, IL-19, IL-20, IL-22, IL-24, and IL-26). A large body of evidence implicates type I interferons in the development of autoimmunity: multiple rheumatic diseases including RA, SLE, systemic sclerosis (SSc), and myositis are characterized by activation of type I interferon signalling28, 29. Mendelian disorders associated with upregulation of type I interferon signalling also present with severe inflammation and autoimmunity30. Paradoxically, though, IFN-β is an approved therapy for MS (as discussed below).
The type II interferon IFN-γ also contributes to the pathogenesis of autoimmune diseases, although the relative contribution of TH1 versus TH17 cells is the subject of ongoing research. TH1 cells, the main producers of IFN-γ, have been implicated in the development of RA and in animal models of collagen-induced arthritis (CIA)11. However, mice lacking the Ifng and Ifngr1 genes are not protected from CIA, and IFN-γ deficiency can even exacerbate the disease. IL-20 is produced by keratinocytes, neutrophils and fibroblast-like synoviocytes31, has been implicated in synoviocyte migration and osteoclast formation, and is upregulated in RA synovium32. Phase 2 clinical trials have shown encouraging results in the treatment of RA32. IL-20 produced by keratinocytes has been implicated in the pathogenesis of psoriatic skin disease31. Other type II cytokines, such as IL-10 and IL-22, have different actions, bringing us to the ‘good guy’ cytokines.
Anti-inflammatory actions of cytokines
Although cytokines are clearly positive effectors of immune responses and drivers of autoimmunity, many type I and II cytokines also have anti-inflammatory actions that need to be considered in the treatment of autoimmune disease. IL-2 is a prime example: the importance of this cytokine in promoting immune tolerance through upregulation of FOXP3 protein would seem to indicate that IL-2 inhibition might induce, rather than treat, autoimmunity,33 but the efficacy of targeting IL-2 in MS provides a very different view9, 10. Whilst daclizumab is efficacious in the treatment of MS, this therapy can be complicated by rash and alopecia, indicative of exacerbation of autoimmunity9, 10, 34.
IL-2 is not alone in having complex actions; many cytokines have both proinflammatory and anti-inflammatory properties. Other cytokines with anti-inflammatory properties include IL-10, IL-22, and IL-35. IL-10 is perhaps the best characterized of these anti-inflammatory cytokines: it suppresses T-cell–dendritic cell/macrophage interactions11, 35 and negatively regulates NLRP3 inflammasome activation to ameliorate synovial inflammation in animal models36. Mutations of IL10 and IL10R cause severe early-onset IBD37. IL-22 is involved in epithelial barrier function and can augment IL-17 function; elevated levels of this cytokine are associated with increased disease activity in RA38. IL-27 can enhance TH1 cell differentiation, but also antagonizes IL-2 production, promotes IL-10 production, and inhibits TH17 cell responses23–25. IL-27 is found at increased levels in the serum and synovial fluid of patients with RA, but whether its role is predominantly proinflammatory or anti-inflammatory is unclear25, 39. However, recent data suggest that IL-27 prevents the development of ectopic lymphoid-like structures in arthritic joints40. Even the interferons, which are crucial for immune responses to viral infections and heavily implicated in the pathogenesis of autoimmune diseases, have immunosuppressive effects; in fact, IFN-β is an approved treatment for MS41. Other “good guy” functions of cytokines include the role of growth factors and hormones that regulate homeostasis and haematopoiesis. Erythropoietin, thrombopoietin, granulocyte colony-stimulating factor (G-CSF), growth hormone, and leptin, among others signal via JAKs42. All of these factors must be considered carefully when designing small-molecule inhibitors of JAKs, which will simultaneously and potently block the actions of multiple factors.
Overview of JAK–STAT signalling
The receptors bound by type I and type II cytokines lack intrinsic catalytic activity; rather, these receptors have an extracellular cytokine-binding domain and a cytoplasmic domain that associates with JAKs. There are four JAKs—JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase 2)—each associates with different cytokine receptors (Table I)8, 42. JAKs are kinases or phosphotransferases, meaning they transfer phosphate from ATP to tyrosine residues of their substrates, an important activation event within cells. One crucial attribute of JAKs is their ability to deposit a phosphate on themselves (autophosphorylation) or on other JAKs (transphosphorylation). Cytokines induce JAK enzymatic activity by binding the extracellular portion of their cognate receptors. This modification enables a variety of signalling molecules to recognize the ‘activated’ receptor.
When one considers the number of cytokines that signal through JAKs, it is not surprising that mutations and polymorphisms of genes encoding these kinases have been linked to diseases of immunity. For example, JAK3 mediates signaling for all the γc cytokines; thus, inactivating mutations of JAK3 recapitulate the phenotype of γc deficiency and cause SCID43, 44. TYK2 loss-of-function causes a milder immunodeficiency45. A large body of evidence from genome-wide association studies has implicated JAKs in the pathogenesis of autoimmune diseases. TYK2 is linked to SLE and Crohn disease46, 47, and JAK2 polymorphisms are associated with Behcet disease48.
STATs comprise an important class of molecules that transmit signals from type I and II cytokine receptors to the nucleus. STATs reside in the cytosol prior to activation. Upon stimulation by cytokines, STATs bind active, phosphorylated receptors and are, in turn, phosphorylated by JAKs. This modification causes STATs to translocate to the nucleus, where they bind DNA and activate the transcription of target genes. There are seven STATS: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6. Like JAKs, the various STATs transmit signals for different cytokine receptors. However, a degree of functional promiscuity exists: in certain situations, one particular STAT protein can transmit signals for cytokines that would usually signal through another STAT protein. Both activating and inactivating mutations of STATs can cause abnormalities in immune function. Loss-of-function mutations STAT1 and STAT2 cause immunodeficiency42. STAT1 gain-of-function mutations lead to chronic mucocutaneous candidiasis via inhibition of IL-17 production42 . Autoimmunity has also been reported in association with STAT1 gain-of-function mutations likely due to increased signaling downstream of IFNγ, IL-21, and IL-649. Dominant negative mutations of STAT3 cause hyper IgE syndrome (also known as Job syndrome), which is characterized by dermatitis, elevated levels of IgE, susceptibility to bacterial pulmonary infection, connective tissue disorders, and bone abnormalities50. Conversely, gain-of-function STAT3 mutations cause early-onset lymphoproliferative disease and autoimmunity51. STAT5 deficiency manifests with immunodeficiency and autoimmunity, as well as dwarfism attributable to the role of STAT5 in mediating growth-hormone signalling52, 53. Genome-wide association studies have established links between STAT3 and Behcet disease48, Crohn’s disease54, and psoriasis54, whereas STAT4 polymorphisms are associated with RA and SLE55. Finally, polymorphisms in STAT6, which mediates IL-4 signalling, are associated with atopy and asthma56. Thus, a large body of evidence points to a critical role for JAKs and STATs in the pathogenesis of rare and common disorders of human immunity.
Jakinibs in autoimmune diseases
The success of ‘biologic therapies’ in treating autoimmunity notwithstanding, the prospect of new alternatives—namely blocking key cytokines by targeting their signal transduction pathways with small molecules—offers a novel and exciting opportunity for treating disease (Figure 2). This possibility was recognized in 199544, but the development of JAK inhibitors (Jakinibs) faced several important challenges. One important consideration is that most Jakinibs act as competitive inhibitors of ATP (Figure 3)42 and all kinases have some degree of homology. Despite such challenges, differences in kinase structure can be capitalized upon to generate inhibitors that exhibit some degree of specificity. First-generation Jakinibs (tofacitinib, ruxolitinib, baricitinib and oclacitinib) inhibit more than one JAK but seem to have reasonable kinome selectivity. Interestingly, none of the first-generation Jakinibs are good TYK2 inhibitors57.
Figure 2.

Type I and II cytokines (red), which are blocked by JAKinibs, are major drivers of autoimmune diseases such as RA. Within the RA joint, FLSs are major sources of TNF, IL-1β, IL-6, and G-CSF. IL-6, along with IL1β, and IL-23, promotes the differentiation of pathogenic TH17 cells. Pathogenic TH17 cells produce the neutrophil growth factor GM-CSF. G-CSF and GM-CSF drive neutrophil-derived production of TNF and IL-1β. TH17 cells are also the major producers of IL-17. IL-17, TNF, and IL-1β induce production of MMPs and ADAMTS, which degrade the cartilage matrix. Similarly, production of the type I cytokine IL-12 by macrophages and DCs promotes the differentiation of pathogenic TH1 cells. TH1 cells produce IFN-γ, activates macrophages. IL-20 and other cytokines promote bone resorption by osteoclasts. DCs also produce IL-15, which promotes FLS survival and TH1 differentiation, and IL-21, which promotes TFH cell differentiation and antibody production.
Abbreviations; ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs; DC, dendritic cell; FLS, fibroblast-like synoviocyte; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocuyte-macrophage colony-stimulating factor; IFN-γ, interferon-γ; MMP, matrix metalloproteinase; TFH cell, follicular helper T cell; TH17 cell, type 17 helper T cell.
Figure 3.

JAKs are composed of several key domains including a tyrosine kinase domain, pseudokinase domain, FERM (band four-point-one, ezrin, radixin, moesin) domain, and an SH2 (Src homology 2) domain. The tyrosine kinase domain binds ATP hydrolyzing it to ADP and catalyzes both autophosphorylation and other substrates including STATs. Jakinibs bind to the pocket ordinarily occupied by ATP, thereby preventing JAKs from using ATP and phosphorylating their substrates.
Tofacitinib
The first Jakinib developed for the treatment of autoimmune diseases was tofacitinib, which inhibits JAK1, JAK3 and, to a lesser extent, JAK258. After promising results in preclinical and early-phase clinical trials for various immune-mediated diseases42, tofacitinib was extensively evaluated in pivotal trials in 2012. Multiple phase 2 and 3 studies demonstrated that tofacitinib was effective for RA as monotherapy or in combination with DMARDs, for treatment-naive or DMARD-refractory disease, and even for disease refractory to treatment with biologic agents59–63. Tofacitinib was superior to methotrexate in the treatment of naive patients64 and as effective as adalimumab63. Two clinical trials assessing MRI and radiographic responses in nearly 1,000 patients indicated that tofacitinib halts structural damage and erosive disease62, 64, 65. Based on these findings, tofacitinib was approved by the US FDA for the treatment of patients with RA with inadequate response to methotrexate.
Tofacitinib has also been found to be effective in the treatment of psoriasis and psoriatic arthritis66, being noninferior to etanercept66–68. However, the FDA has declined to approve tofacitinib for psoriasis. This may be because higher doses of tofacitinib (10 mg twice daily) than the approved dose approved for RA (5 mg twice daily) were required optimal efficacy. The higher dose (10 mg twice daily) was not approved by the FDA because of concerns regarding adverse events69.. Studies also indicate that continuous therapy results in more durable remissions, although up to 60% of patients can recapture a response when treated intermittently70. Tofacitinib is also an efficacious treatment for ulcerative colitis71 and possibly for Crohn disease, although results in the latter are inconsistent72. In prophylaxis of kidney transplant rejection, tofacitinib preserves renal function better than standard-of-care with calcineurin inhibitors, by preventing tubular atrophy and interstitial fibrosis73.
Ruxolitinib and baricitinib
The JAK1 and JAK2 inhibitor ruxolitinib was actually the first FDA-approved Jakinib, for the treatment of myeloproliferative disease42. However, ruxolitinib seems to be efficacious in models of preclinical arthritis74 and results of a phase II trial in RA are promising (ClinicalTrials.gov identifier NCT00550043).21
Another JAK1 and JAK2 inhibitor, baricitinib, has shown efficacy in the treatment of RA, in which setting—like tofacitinib—it is effective in patients with disease refractory to treatment with DMARDs and TNF inhibitors75–78. Baricitinib halts radiographic disease progression, with significant differences in erosions and joint-space narrowing, particularly at the higher dose of 4 mg twice daily, and to a lesser degree with 2 mg twice daily.75–78
Oclacitinib
Oclacitinib is a nonselective Jakinib approved for the treatment of eczema in dogs. Orally administered Jakinibs are not currently being tested for the treatment of allergic diseases; however, the efficacy of oclacitinib in canine atopic dermatitis79 demonstrates that inhibition of JAKs can disrupt key signalling pathways that drive allergic inflammation80. These findings have paved the way for clinical trials of topical tofacitinib for the treatment of atopic skin disease in humans (ClinicalTrials.gov identifier NCT02001181).21
Adverse effects of Jakinibs
As with other effective therapies in autoimmune disease, Jakinibs are not without side effects. Many of the adverse effects of Jakinibs can, like their therapeutic efficacy, be directly traced to their mechanism of action: blocking JAK-dependent cytokine signalling. Because more patients have been treated with tofacitinib than other agents, we know the most about this drug; however, the adverse effect profile of baricitinib seems comparable75–78.
Infection
An obvious concern with the use of any immunomodulatory drug is the risk of infection, and in clinical trials increased rates of infections have been demonstrated in tofacitinib-treated patients. These infections were mainly upper respiratory tract infections, urinary tract infections, and viral gastroenteritis. However, serious cases of tuberculosis, fungal infection, Pneumocystis jirovecii pneumonia, and bacterial pneumonia were reported as well66, 73, 81–84. Given the broad range of cytokines that can be affected by tofacitinib, this outcome was not unexpected.
The risk of infection with tofacitinib use is comparable to that with other biologic agents83, 84, with one exception: the risk of varicella zoster virus (VZV) infection seems to be increased83, 85. This increase could be attributable to the effects of tofacitinib on interferon or IL-15 signalling, the latter being a key cytokine for NK-cell homeostasis86. However, NK cell counts are only moderately reduced in tofacitinib-treated patients and do not correlate with infection risk87, 87, 88. Tofacitinib-treated patients also have impaired responses to vaccination89, so clinicians are encouraged to immunize patients against pneumococcus and VZV prior to initiating this therapy, as is also recommended with other biologic agents. In renal transplant recipients, higher rates of BK virus infection were seen in those treated with tofacitinib than with ciclosporin, although the drugs were used in the setting of combination immunosuppressive therapy90. Whilst most of these infections do not result in nephropathy, a trend towards higher rates of BK virus-associated nephropathy in tofacitinib-treated patients was observed.90 Clinical trials and long-term extension studies have not revealed similar issues in patients with RA83, 84, but clearly this potential risk must be monitored as new Jakinibs emerge for various clinical indications and as part of combined treatment regimens.
Lipid effects
Increases in levels of total cholesterol, LDL cholesterol, and HDL cholesterol have been noted in clinical trials of tofacitinib42, 83, 84 This side effect is also seen with IL-6 receptor (IL-6R) blocker tocilizumab91,92 IL-6 promotes insulin resistance and redistributes fatty acids from the blood to peripheral tissues, thereby decreasing serum lipids92, 93. Thus, treatment-induced changes in lipid levels may or may not correlate with rates of cardiovascular disease. The results of clinical trials and long-term extension data do not indicate an increased risk of cardiovascular disease with tofacitinib use83, 93. On the contrary, tofacitinib can improve arterial stiffness, suggesting that it might improve cardiovascular outcomes in patients with RA94. In this context, tofacitinib-induced hypercholesterolemia could be cardioprotective. Indeed, patients with RA commonly have low total, HDL, and LDL cholesterol levels and a discordantly increased risk of cardiovascular disease93. Moreover, Jakinibs alter lipoprotein kinetics95 and increase LDL particle size95,95. Jakinibs also confer metabolic properties of brown fat to white adipocytes by uncoupling the respiratory chain and increasing mitochondrial activity96, a novel and provocative mechanism by which JAK blockade might reduce cardiovascular risk. Needless to say, the metabolic effects of JAK blockade are complex and deserve further attention.
Other Effects
Because all first-generation Jakinibs inhibit JAK2, which mediates erythropoietin, thrombopoietin, IL-11, G-CSF and GM-CSF signalling, patients treated with these agents can develop cytopaenias, especially anaemia; generally, this effect is not necessarily a cause for discontinuation of treatment. Other adverse effects reported in clinical trials include minimal elevations in serum transaminases42 and reductions in glomerular filtration rate81. Neither of these laboratory abnormalities seems to indicate any substantial end-organ damage, with the possible exception of BK nephropathy83.
Other considerations
Although the adverse events attributed to Jakinibs in clinical trials for rheumatic diseases indicate an acceptable safety profile, several additional factors warrant consideration. Cytokines such as IL-10, IL-22 and IL-27 have important anti-inflammatory actions; other cytokines such as IL-2 have both inflammatory and immunosuppressive actions. Whether suppression of these anti-inflammatory actions will be clinically relevant in the context of inflammatory cytokine blockade remains to be determined.
Finally, the potential role of JAK inhibition in tumour development warrants discussion. Because interferons, IL-12, and other cytokines contribute to the elimination and destruction of tumours, prolonged blockade of JAK–STAT signalling might promote malignancy97. However, as is the case with biologic therapies, whether Jakinib-treated patients are truly at increased risk of cancer is unclear. In RA trials, the risk of malignancy in tofacitinib-treated patients was not significantly increased83. Although tofacitinib is associated with an increased risk of nonmelanoma skin cancer, the effect size is similar to that seen with other biologic agents such as TNF inhibitors82. When Jakinibs have been used for transplant-rejection prophylaxis in combination with other immunosuppressive drugs, the risk of developing post-transplant lymphoproliferative disease (PTLD) is increased.
Other uses of Jakinibs
The future of therapies that target JAKs and the cytokines that signal through them is only starting to emerge. One need only consider the example of tocilizumab to see the potential applicability of JAK blockade to a wide variety of immune-mediated diseases. Initially used for the treatment of RA, tocilizumab is now approved for use in polyarticular juvenile idiopathic arthritis (JIA) and systemic-onset JIA98, 99 and has been used successfully to treat large-vessel vasculitis100, polymyalgia rheumatica101, AA amyloidosis102, spondyloarthritis103, SSc104, and myositis105, among other rheumatic diseases. As IL-6 is clearly blocked by existing Jakinibs, these diseases should be reasonable settings in which to test JAK blockade.
Alopecia areata is characterized by an IFN-γ and γc cytokine gene signature89, or upregulation of these genes. Tofacitinib, ruxolitinib, and baricitinib have all been used successfully to treat alopecia areata and alopecia universalis106–109, and clinical trials in these diseases are ongoing (ClinicalTrials.gov identifiers NCT02299297, NCT02197455, NCT02312882, NCT01950780)21. Preliminary evidence indicates that tofacitinib could be equally effective in the treatment of vitiligo110, and that inhibition of JAK-STAT signaling causes hair growth through initiation of anagen111.
The presence of the type I inteferon gene signature in SLE, myositis, primary Sjogren’s syndrome, and SSc, and perhaps a subset of RA, suggests that Jakinibs might be useful in these settings28, 29, 112, 113. A case of refractory dermatomyositis was indeed reported to respond to treatment with ruxolitinib114. The potential for Jakinibs to treat interferon-mediated diseases has also been suggested by preliminary data in patients with interferonopathies—monogenic diseases characterized by uncontrolled upregulation of type I intereferons115. A ‘compassionate use’ study of baricitinib in chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) and STING-associated vasculopathy with onset in infancy (SAVI) is ongoing116 (ClinicalTrials.gov identifier NCT01724580)21.
An important consideration involves the optimal dosing and route of administration for Jakinibs. Although the current paradigm of continuous low-level dosing is comparable to that of DMARDs and other maintenance therapies, it is possible that Jakinibs might be most effective when used in high doses to induce remission followed by tapering to much lower doses. Moreover, topical formulations of tofacitinib improve symptoms of dry eye disease117 and are being investigated for psoriasis and eczema (ClinicalTrials.gov identifiers NCT01831466, NCT02193815, NCT02001181, and NCT00678561)21. If these formulations prove as efficacious as systemic therapy, they could fundamentally alter the treatment of diseases that affect a single organ system—including pulmonary and gastrointestinal diseases.
Jakinibs: the next generation
The assessment of the adverse events associated with tofacitinib led the European Medical Agency to refuse marketing approval for tofacitinib for the treatment of RA. Other agencies had a different view of the relative risks and benefits of this drug. As with any immunomodulatory drug, the goal is to identify regimens that provide efficacy at the minimal effective dose. As more experience is gained with tofacitinib and other Jakinibs in a range of disorders, we should have a better understanding of the risks and benefits of these agents and perhaps more insights into optimal dosing and regimens in real-world usage. Whether the assessment of the relative benefits and risks of baricitinib and other agents in development will or will not be viewed similarly to those of tofacitinib remains to be seen.
However, with the demonstrated efficacy of multiple first-generation Jakinibs that exhibit kinome selectivity but target multiple JAKs, the question naturally arises of whether targeting a single JAK would be superior. Might such agents exhibit similar efficacy with fewer adverse events? Conversely though, agents that block fewer JAKs by definition block fewer inflammatory cytokines, which could negatively affect their therapeutic efficacy. Indeed, development of second-generation Jakinibs with a greater degree of JAK selectivity is underway. Phase 1 and 2 trials seem to indicate that the JAK1 inhibitor filgotinib118 and the JAK3 inhibitor decernotinib119, 120 are effective for the treatment of RA. Several important questions will need to be answered as these and other selective Jakinibs emerge on the clinical scene. The apparent selectivity of these agents needs to be verified in vivo, by examining lymphoid, myeloid, and erythroid compartments for signs that other JAKs—especially JAK2—are being affected. Furthermore, even if an inhibitor is exquisitely selective for one JAK over another, selectivity is not necessarily ensured across the kinome. This was the case for a recently developed class of reversible covalent JAK3 inhibitors that also targeted several other tyrosine kinases121. At present, ~20 Jakinibs are in clinical trials for various autoimmune conditions including autoinflammatory disease, RA, psoriasis and psoriatic arthritis, SLE, JIA, spondyloarthritis, discoid lupus, ulcerative colitis, atopic dermatitis, and diabetic nephropathy (ClinicalTrials.gov identifiers NCT01458574, NCT01724580, NCT01687309, NCT01500551, NCT01597050, and NCT02001181)21.
Targeting STATs?
In principle, cytokine signalling could also by blocked by inhibiting STAT activation, interrupting STAT–receptor interactions, blocking STAT dimerization, or interfering with STAT–DNA binding42, 122. These strategies have been considered especially in cancer, in which constitutive JAK–STAT activation is a common event42. However, unlike JAKs, STATs are not enzymes, and generating drug candidates suitable for clinical use has been difficult owing to challenges of bioavailability, in vivo efficacy, and selectivity; for instance, STAT3, a heavily investigated target in the treatment of solid-organ malignancy, has a high degree of homology with STAT1. Moreover, STATs represent convergent endpoints for multiple signalling pathways with critical roles in tumour prevention and host defense123. These issues must be considered if STATs are to be targeted in the treatment of rheumatic diseases.
Conclusions
Cytokines that signal through JAKs and STATs are fundamental drivers of host defense, immunity, and inflammation. Advances in the understanding of cytokine biology, and of the links between cytokine regulation and autoimmunity, have sparked a revolution in the treatment of rheumatic diseases over the past 15 years. However, the continuing burden of immune-mediated diseases clearly demonstrates that novel forms of therapy are needed. The emergence of small-molecule targeted therapies represents a new phase in targeted therapy owing to the ability of these agents to simultaneously block multiple signalling pathways. Tofacitinib is the first targeted small molecule to be approved for use in rheumatologic diseases and is likely to be joined by a number of other Jakinibs. A new chapter in the treatment of immune-mediated diseases has begun, in which the paradigm has been fundamentally shifted yet again. The next 5 years will be a crucial time, as we learn how to best use Jakinibs and other small-molecule inhibitors for the treatment of the vast array of autoimmune diseases encountered by clinicians.
Table II.
JAKs and disease
| JAK | Human disease: gain of function | Human disease: loss of function |
|---|---|---|
| JAK1 | ALL, AML, solid organ malignancies | Not applicable |
| JAK2 | PCV*, MF*, ET*, hypercoagulable state, somatic mutations associated with acute and chronic hematologic malignancies | Not applicable |
| JAK3 | Acute megakaryoblastic leukemia, T cell leukemias and lymphomas | Severe combined immunodeficiency (SCID), Jacobsen syndrome |
| TYK2 | Cutaneous lymphoproliferative disorders, T cell leukemias | Primary immunodeficiency |
PCV = polycythemia vera, MF = myelofibrosis, ET = essential thrombocythemia
Table II describes known associations of JAK mutations with human disease. Rare JAK mutations are associated with disorders of immune function, and JAKs can be activated in a variety of cancers
Table III.
STATs and disease
| STAT | Human disease: gain of function | Human disease: loss of function | Human disease: polymorphism |
|---|---|---|---|
| STAT1 | Chronic mucocutaneous candidiasis | Susceptibility to mycobacterial and viral infections | Not applicable |
| STAT2 | Not applicable | Increased susceptibility to viral infections | Not applicable |
| STAT3 | Lymphoproliferative disease and autoimmunity, Large granulocytic leukemia (LGL) | Autosomal-dominant Hyper IgE (Job’s) syndrome | Behcet’s disease |
| STAT4 | Not applicable | Not applicable | RA, SLE |
| STAT5a/STAT5b | Somatic mutations associated with LGL | Autoimmunity, bleeding diathesis, immunodeficiency and dwarfism | Not applicable |
| STAT6 | Not applicable | Not applicable | Asthma, atopy, high serum IgE |
Table III describes known associations of STAT mutations with human disease. Rare STAT mutations are associated with disorders of immune function, and STATs are commonly activated in a variety of cancers
Table IV.
Jakinibs being investigated for rheumatic diseases
| Drug | Target | Status | Diseases |
|---|---|---|---|
| Ruxolitinib (INC424) | JAK1, JAK2 |
FDA approved Phase II Phase IIb |
RA Various cancers Psoriasis (topical) |
| Tofacitinib (CP690550) | JAK3>JAK1>> (JAK2) |
FDA approved Phase III Phase II |
RA Psoriasis, Ulcerative colitis Spondyloarthritis, JIA Transplant rejection |
| Oclacitinib (PF03394197) | JAK1 | FDA approved | Canine allergic dermatitis |
| ABT494 | JAK1 | Phase II | RA, Crohn’s |
| Baricitinib (INCB28050, LY3009104) | JAK1, JAK2 | Phase II | RA, Psoriasis, Diabetic nephropathy, autoinflammatory disease |
| Filgotinib (GLPG0634) | JAK1 | Phase II | RA, Crohn’s disease |
| INCB039110 | JAK1, JAK2 | Phase II | Psoriasis, RA |
| Peficitinib (ASP015K) | pan-JAK | Phase II | Psoriasis, RA |
| R333 | JAK/SYK | Phase II | Discoid lupus (topical) |
| GLG0778 | JAK1 | Phase II | SLE |
| GSK2586184 | JAK1 | Phase II | SLE, Psoriasis |
| Decernotinib (VX509) | JAK3 | Phase IIb | RA |
Table IV denotes JAKinibs that are FDA-approved for, or in clinical trials for, the treatment of autoimmune diseases.
Abbreviations: RA = rheumatoid arthritis, JIA = juvenile idiopathic arthritis, SYK = spleen tyrosine kinase
Key points.
Cytokines are major drivers of autoimmunity, and targeting cytokines has revolutionized the treatment of rheumatologic diseases
-
-
Despite their success, biologic agents that target key cytokines are not completely effective in all patients
-
-
Type I and II cytokines signal through the JAK–STAT pathway, and pharmacological inhibition of this signal transduction pathway with small molecules can block the actions of these cytokines
-
-
JAK inhibitors, or Jakinibs, are effective for rheumatoid arthritis and other immune-mediated diseases
-
-
Many of the adverse effects of Jakinibs can be linked to action of the cytokines that are blocked
-
-
Jakinibs are currently being investigated for a number of new indications, and second-generation selective Jakinibs are being developed and tested
Acknowledgments
The work of D.M.S., M.G. and JOS. is supported by the Intramural Research Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the NIH.
Biographies
Daniella Muallem Schwartz
Daniella Schwartz graduated from Rice University. She obtained her MD from Wake Forest University School of Medicine, NC, USA, and then trained in Internal Medicine at Virginia Commonwealth University, VA, USA. She completed her rheumatology fellowship at the NIH in 2015 and is currently a Metzger Scholar in Translational Research. She is interested in the epigenetic factors that determine T cell lineage commitment, and the role of these factors in driving autoimmunity.
John Joseph O’Shea
John J. O’Shea graduated Phi Beta Kappa from St Lawrence University with a BSc degree, and then gained a Doctor of Medicine degree from the University of Cincinnati. He completed a residency in Internal Medicine at the State University of New York Upstate Medical University, subspecialty training at the National Institute of Allergy and Infectious Diseases, NIH, and further postdoctoral training at the National Institute of Child Health and Human Development. He is currently the Director of the Intramural Research Program at the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH. Dr. O’Shea served as the Acting Director of the NIH Center for Regenerative Medicine 2009–2011. Dr O’Shea cloned JAK3 and his interest for the past 20 years has been cytokine signalling. He is a member of the National Academy of Medicine and has won numerous prizes including the Ross Prize in Molecular Medicine.
Michael Bonelli
Dr Michael Bonelli obtained his MD from the Medical University of Vienna, Austria. He finished his training in Internal Medicine at the general hospital of Vienna, where he is currently a Rheumatology fellow. He started his scientific career at the Department of Rheumatology at the Medical University of Vienna, where he investigated the role of regulatory T cells in patients with systemic lupus erythematosus. Between 2011 and 2015 he worked in the laboratory of Dr John O’Shea, where he analyzed the plasticity of regulatory T cells under inflammatory conditions. Currently, he is working at the Medical University of Vienna exploring the role of T cells in inflammatory and autoimmune diseases.
Massimo Gadina
Dr Massimo Gadina received his PhD in medicinal chemistry and technology at the Universitá di Milano, Italy, and MD from the Université de Dijon, Dijon, France. He is currently the Director of the Office of Science and Technology at NIAMS. Prior to this appointment, he was Senior Lecturer at the Division of Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, The Queen’s University of Belfast, Northern Ireland, UK. Dr Gadina’s research interests are focused on immune-mediated diseases and specifically the biology of cytokines and their relative signalling pathways. His translational work is also focused on autoinflammation and other inflammatory diseases.
Footnotes
Competing interests
J.OS. declares that he and the US Government receive royalties based on patents related to targeting Janus kinases. JOS and M.G. and the US Government have had longstanding Cooperative Research and Development Agreements with Pfizer, which produces tofacitinib, a Janus kinase inhibitor.
Author contributions
D.M.S., M.B., M.G. and JOS researched the data, provided a substantial contribution to discussions of the content, contributed to writing the article, and reviewed/edited the manuscript before submission.
References
- 1.McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365:2205–19. doi: 10.1056/NEJMra1004965. [DOI] [PubMed] [Google Scholar]
- 2.Lubberts E. The IL-23-IL-17 axis in inflammatory arthritis. Nat Rev Rheumatol. 2015 doi: 10.1038/nrrheum.2015.128. [DOI] [PubMed] [Google Scholar]
- 3.O’Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev Immunol. 2002;2:37–45. doi: 10.1038/nri702. [DOI] [PubMed] [Google Scholar]
- 4.Bell E. Inflammation: Targeting TNF. Nature Reviews Immunology. 2009;9:390–391. [Google Scholar]
- 5.Scott DL. Biologics-based therapy for the treatment of rheumatoid arthritis. Clin Pharmacol Ther. 2012;91:30–43. doi: 10.1038/clpt.2011.278. [DOI] [PubMed] [Google Scholar]
- 6.O’Shea JJ, Gadina M, Schreiber RD. Cytokine Signaling in 2002. Cell. 2002;109:S121–S131. doi: 10.1016/s0092-8674(02)00701-8. [DOI] [PubMed] [Google Scholar]
- 7.Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
- 8.O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med. 2013;368:161–70. doi: 10.1056/NEJMra1202117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Giovannoni G, et al. Daclizumab high-yield process in relapsing-remitting multiple sclerosis (SELECTION): a multicentre, randomised, double-blind extension trial. The Lancet Neurology. 2014;13:472–481. doi: 10.1016/S1474-4422(14)70039-0. [DOI] [PubMed] [Google Scholar]
- 10.Gold R, et al. Daclizumab high-yield process in relapsing-remitting multiple sclerosis (SELECT): a randomised, double-blind, placebo-controlled trial. The Lancet. 2013;381:2167–2175. doi: 10.1016/S0140-6736(12)62190-4. [DOI] [PubMed] [Google Scholar]
- 11.McInnes IB, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol. 2007;7:429–42. doi: 10.1038/nri2094. [DOI] [PubMed] [Google Scholar]
- 12.Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol. 2009;9:480–90. doi: 10.1038/nri2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Spits H, Di Santo JP. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat Immunol. 2011;12:21–7. doi: 10.1038/ni.1962. [DOI] [PubMed] [Google Scholar]
- 14.Kaplan MH, Hufford MM, Olson MR. The development and in vivo function of T helper 9 cells. Nat Rev Immunol. 2015;15:295–307. doi: 10.1038/nri3824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Eto D, et al. IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One. 2011;6:e17739. doi: 10.1371/journal.pone.0017739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kotlarz D, Zietara N, Milner JD, Klein C. Human IL-21 and IL-21R deficiencies: two novel entities of primary immunodeficiency. Curr Opin Pediatr. 2014;26:704–12. doi: 10.1097/MOP.0000000000000160. [DOI] [PubMed] [Google Scholar]
- 17.Rasmussen TK, et al. Increased interleukin 21 (IL-21) and IL-23 are associated with increased disease activity and with radiographic status in patients with early rheumatoid arthritis. J Rheumatol. 2010;37:2014–20. doi: 10.3899/jrheum.100259. [DOI] [PubMed] [Google Scholar]
- 18.Spolski R, Leonard WJ. Interleukin-21: a double-edged sword with therapeutic potential. Nat Rev Drug Discov. 2014;13:379–95. doi: 10.1038/nrd4296. [DOI] [PubMed] [Google Scholar]
- 19.Papatriantafyllou M. Cytokines: GM-CSF in focus. Nat Rev Immunol. 2011;11:370–1. doi: 10.1038/nri2996. [DOI] [PubMed] [Google Scholar]
- 20.Burmester GR, et al. Efficacy and safety of mavrilimumab in subjects with rheumatoid arthritis. Ann Rheum Dis. 2013;72:1445–52. doi: 10.1136/annrheumdis-2012-202450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.(US National Library of Medicine, 2015)
- 22.Cornelissen C, Luscher-Firzlaff J, Baron JM, Luscher B. Signaling by IL-31 and functional consequences. Eur J Cell Biol. 2012;91:552–66. doi: 10.1016/j.ejcb.2011.07.006. [DOI] [PubMed] [Google Scholar]
- 23.Hirahara K, et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity. 2012;36:1017–30. doi: 10.1016/j.immuni.2012.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hirahara K, et al. Mechanisms underlying helper T-cell plasticity: implications for immune-mediated disease. J Allergy Clin Immunol. 2013;131:1276–87. doi: 10.1016/j.jaci.2013.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hunter CA, Kastelein R. Interleukin-27: balancing protective and pathological immunity. Immunity. 2012;37:960–9. doi: 10.1016/j.immuni.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Griffiths CE, et al. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med. 2010;362:118–28. doi: 10.1056/NEJMoa0810652. [DOI] [PubMed] [Google Scholar]
- 27.Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol. 2014;14:329–42. doi: 10.1038/nri3661. [DOI] [PubMed] [Google Scholar]
- 28.Higgs BW, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70:2029–36. doi: 10.1136/ard.2011.150326. [DOI] [PubMed] [Google Scholar]
- 29.Ronnblom L, Eloranta ML. The interferon signature in autoimmune diseases. Curr Opin Rheumatol. 2013;25:248–53. doi: 10.1097/BOR.0b013e32835c7e32. [DOI] [PubMed] [Google Scholar]
- 30.Crow YJ. Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci. 2011;1238:91–8. doi: 10.1111/j.1749-6632.2011.06220.x. [DOI] [PubMed] [Google Scholar]
- 31.Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines–from host defence to tissue homeostasis. Nat Rev Immunol. 2014;14:783–95. doi: 10.1038/nri3766. [DOI] [PubMed] [Google Scholar]
- 32.Senolt L, et al. Efficacy and Safety of Anti-Interleukin-20 Monoclonal Antibody in Patients With Rheumatoid Arthritis: A Randomized Phase IIa Trial. Arthritis Rheumatol. 2015;67:1438–48. doi: 10.1002/art.39083. [DOI] [PubMed] [Google Scholar]
- 33.Huss DJ, et al. In vivo maintenance of human regulatory T cells during CD25 blockade. J Immunol. 2015;194:84–92. doi: 10.4049/jimmunol.1402140. [DOI] [PubMed] [Google Scholar]
- 34.Oh J, et al. Daclizumab-induced adverse events in multiple organ systems in multiple sclerosis. Neurology. 2014;82:984–8. doi: 10.1212/WNL.0000000000000222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ouyang W, Rutz S, Crellin NK, Valdez PA, Hymowitz SG. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011;29:71–109. doi: 10.1146/annurev-immunol-031210-101312. [DOI] [PubMed] [Google Scholar]
- 36.Greenhill CJ, et al. Interleukin-10 regulates the inflammasome-driven augmentation of inflammatory arthritis and joint destruction. Arthritis Res Ther. 2014;16:419. doi: 10.1186/s13075-014-0419-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Glocker EO, Kotlarz D, Klein C, Shah N, Grimbacher B. IL-10 and IL-10 receptor defects in humans. Ann N Y Acad Sci. 2011;1246:102–7. doi: 10.1111/j.1749-6632.2011.06339.x. [DOI] [PubMed] [Google Scholar]
- 38.da Rocha LF, Jr, et al. Increased serum interleukin 22 in patients with rheumatoid arthritis and correlation with disease activity. J Rheumatol. 2012;39:1320–5. doi: 10.3899/jrheum.111027. [DOI] [PubMed] [Google Scholar]
- 39.Shen H, Xia L, Xiao W, Lu J. Increased levels of interleukin-27 in patients with rheumatoid arthritis. Arthritis Rheum. 2011;63:860–1. doi: 10.1002/art.30180. [DOI] [PubMed] [Google Scholar]
- 40.Jones GW, et al. Interleukin-27 inhibits ectopic lymphoid-like structure development in early inflammatory arthritis. J Exp Med. 2015;212:1793–802. doi: 10.1084/jem.20132307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gonzalez-Navajas JM, Lee J, David M, Raz E. Immunomodulatory functions of type I interferons. Nat Rev Immunol. 2012;12:125–35. doi: 10.1038/nri3133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.O’Shea JJ, et al. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–28. doi: 10.1146/annurev-med-051113-024537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Macchi P, et al. Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID) Nature. 1995;377:65–8. doi: 10.1038/377065a0. [DOI] [PubMed] [Google Scholar]
- 44.Russell SM, et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science. 1995;270:797–800. doi: 10.1126/science.270.5237.797. [DOI] [PubMed] [Google Scholar]
- 45.Minegishi Y, et al. Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple cytokine signals involved in innate and acquired immunity. Immunity. 2006;25:745–55. doi: 10.1016/j.immuni.2006.09.009. [DOI] [PubMed] [Google Scholar]
- 46.Sigurdsson S, et al. Polymorphisms in the tyrosine kinase 2 and interferon regulatory factor 5 genes are associated with systemic lupus erythematosus. Am J Hum Genet. 2005;76:528–37. doi: 10.1086/428480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Franke A, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118–25. doi: 10.1038/ng.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hu K, Hou S, Jiang Z, Kijlstra A, Yang P. JAK2 and STAT3 polymorphisms in a Han Chinese population with Behcet’s disease. Invest Ophthalmol Vis Sci. 2012;53:538–41. doi: 10.1167/iovs.11-8440. [DOI] [PubMed] [Google Scholar]
- 49.Uzel G, et al. Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation-polyendocrinopathy-enteropathy-X-linked-like syndrome. J Allergy Clin Immunol. 2013;131:1611–23. doi: 10.1016/j.jaci.2012.11.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Holland SM, et al. STAT3Mutations in the Hyper-IgE Syndrome. New England Journal of Medicine. 2007;357:1608–1619. doi: 10.1056/NEJMoa073687. [DOI] [PubMed] [Google Scholar]
- 51.Milner JD, et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood. 2015;125:591–9. doi: 10.1182/blood-2014-09-602763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Cohen AC, et al. Cutting Edge: Decreased Accumulation and Regulatory Function of CD4+CD25high T Cells in Human STAT5b Deficiency. The Journal of Immunology. 2006;177:2770–2774. doi: 10.4049/jimmunol.177.5.2770. [DOI] [PubMed] [Google Scholar]
- 53.Kofoed EM, et al. Growth hormone insensitivity associated with a STAT5b mutation. N Engl J Med. 2003;349:1139–47. doi: 10.1056/NEJMoa022926. [DOI] [PubMed] [Google Scholar]
- 54.Ellinghaus D, et al. Combined analysis of genome-wide association studies for Crohn disease and psoriasis identifies seven shared susceptibility loci. Am J Hum Genet. 2012;90:636–47. doi: 10.1016/j.ajhg.2012.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Remmers EF, et al. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 2007;357:977–86. doi: 10.1056/NEJMoa073003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Duetsch G, et al. STAT6 as an asthma candidate gene: polymorphism-screening, association and haplotype analysis in a Caucasian sib-pair study. Hum Mol Genet. 2002;11:613–21. doi: 10.1093/hmg/11.6.613. [DOI] [PubMed] [Google Scholar]
- 57.O’Shea JJ, Kontzias A, Yamaoka K, Tanaka Y, Laurence A. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis. 2013;72(Suppl 2):ii111–5. doi: 10.1136/annrheumdis-2012-202576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Changelian PS, et al. Prevention of organ allograft rejection by a specific Janus kinase 3 inhibitor. Science. 2003;302:875–8. doi: 10.1126/science.1087061. [DOI] [PubMed] [Google Scholar]
- 59.Fleischmann R, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. 2012;367:495–507. doi: 10.1056/NEJMoa1109071. [DOI] [PubMed] [Google Scholar]
- 60.Lee EB, Fleischmann R, Hall S. Radiographic, clinical and functional comparison of tofacitinib monotherapy versus methotrexate in methotrexate-naïve patients with rheumatoid arthritis. Arthritis Rheum. 2012;64:S1049. [Google Scholar]
- 61.Burmester GR, et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. The Lancet. 2013;381:451–460. doi: 10.1016/S0140-6736(12)61424-X. [DOI] [PubMed] [Google Scholar]
- 62.van der Heijde D, et al. Tofacitinib (CP-690,550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic study. Arthritis Rheum. 2013;65:559–70. doi: 10.1002/art.37816. [DOI] [PubMed] [Google Scholar]
- 63.van Vollenhoven RF, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367:508–19. doi: 10.1056/NEJMoa1112072. [DOI] [PubMed] [Google Scholar]
- 64.Lee EB, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med. 2014;370:2377–86. doi: 10.1056/NEJMoa1310476. [DOI] [PubMed] [Google Scholar]
- 65.Conaghan PG, et al. Effects of Tofacitinib on MRI enpoints in Methotrexate-Naive Early Rheumatoid Arthritis: A Phase 2 MRI Study with Semi-quantitative and Quantitative Endpoints. Annals of the Rheumatic Diseases. 2015 [Google Scholar]
- 66.Chiricozzi A, et al. Tofacitinib for the treatment of moderate-to-severe psoriasis. Expert Rev Clin Immunol. 2015;11:443–55. doi: 10.1586/1744666X.2015.1013534. [DOI] [PubMed] [Google Scholar]
- 67.Bachelez H, et al. Tofacitinib versus etanercept or placebo in moderate-to-severe chronic plaque psoriasis: a phase 3 randomised non-inferiority trial. The Lancet. 2015 doi: 10.1016/S0140-6736(14)62113-9. [DOI] [PubMed] [Google Scholar]
- 68.Pfizer. in Pfizer Press Release (http://www.pfizer.com/news/press-release/press-release-detail/pfizer_announces_oral_tofacitinib_meets_primary_endpoints_in_pivotal_phase_3_psoriasis_trials, 2015)
- 69.Pfizer. in Pfizer Press Release (http://www.pfizer.com/news/press-release/press-release-detail/pfizer_receives_complete_response_letter_from_fda_for_oral_xeljanz_tofacitinib_citrate_supplemental_new_drug_application_for_moderate_to_severe_chronic_plaque_psoriasis, 2015)
- 70.Bissonnette R, et al. Tofacitinib withdrawal and retreatment in moderate-to-severe chronic plaque psoriasis: a randomized controlled trial. Br J Dermatol. 2015;172:1395–406. doi: 10.1111/bjd.13551. [DOI] [PubMed] [Google Scholar]
- 71.Sandborn WJ, et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med. 2012;367:616–24. doi: 10.1056/NEJMoa1112168. [DOI] [PubMed] [Google Scholar]
- 72.Sandborn WJ, et al. A Phase 2 Study of Tofacitinib, an Oral Janus Kinase Inhibitor, in Patients With Crohn’s Disease. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2014.01.029. [DOI] [PubMed] [Google Scholar]
- 73.Vincenti F, et al. Evaluation of the effect of tofacitinib exposure on outcomes in kidney transplant patients. Am J Transplant. 2015;15:1644–53. doi: 10.1111/ajt.13181. [DOI] [PubMed] [Google Scholar]
- 74.Fridman JS, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol. 2010;184:5298–307. doi: 10.4049/jimmunol.0902819. [DOI] [PubMed] [Google Scholar]
- 75.Keystone EC, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis. 2015;74:333–40. doi: 10.1136/annrheumdis-2014-206478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Smolen JS, et al. Patient-Reported Outcomes from a Phase 3 Study of Baricitinib in Patients with Rheumatoid Arthritis (RA) and an Inadequate Response to Tumor Necrosis Factor Inhibitors. Annals of the Rheumatic Diseases. 2015 doi: 10.1136/annrheumdis-2016-209821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Genovese MC, et al. Baricitinib, an Oral Janus Kinase (JAK)1/JAK2 Inhibitor, in Patients with Active Rheumatoid Arthritis (RA) and an Inadequate Response to TNF Inhibitors: Results of the Phase 3 RA-BEACON Study. Annals of the Rheumatic Diseases. 2015 [Google Scholar]
- 78.Dougados M, et al. Baricitinib, an Oral Janus Kinase (JAK)1/JAK2 Inhibitor, in Patients with Active Rheumatoid Arthritis (RA) and an Inadequate Response to cDMARD Therapy: Results of the Phase 3 RA-BUILD Study. Annals of the Rheumatic Diseases. 2015 [Google Scholar]
- 79.Cosgrove SB, et al. A blinded, randomized, placebo-controlled trial of the efficacy and safety of the Janus kinase inhibitor oclacitinib (Apoquel(R)) in client-owned dogs with atopic dermatitis. Vet Dermatol. 2013;24:587–97. e141–2. doi: 10.1111/vde.12088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Gonzales AJ, et al. Oclacitinib (APOQUEL((R))) is a novel Janus kinase inhibitor with activity against cytokines involved in allergy. J Vet Pharmacol Ther. 2014;37:317–24. doi: 10.1111/jvp.12101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kremer JM, et al. Evaluation of the effect of tofacitinib on measured glomerular filtration rate in patients with active rheumatoid arthritis: results from a randomised controlled trial. Arthritis Res Ther. 2015;17:95. doi: 10.1186/s13075-015-0612-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Curtis J, et al. Analysis of Non-Melanoma Skin Cancer Across the Tofacitinib Rheumatoid Arthritis Clinical Programme. Annals of the Rheumatic Diseases. 2015 [PubMed] [Google Scholar]
- 83.Wollenhaupt J, et al. Safety and efficacy of tofacitinib, an oral janus kinase inhibitor, for the treatment of rheumatoid arthritis in open-label, longterm extension studies. J Rheumatol. 2014;41:837–52. doi: 10.3899/jrheum.130683. [DOI] [PubMed] [Google Scholar]
- 84.He Y, et al. Efficacy and safety of tofacitinib in the treatment of rheumatoid arthritis: a systematic review and meta-analysis. BMC Musculoskelet Disord. 2013;14:298. doi: 10.1186/1471-2474-14-298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Winthrop KL, et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014;66:2675–84. doi: 10.1002/art.38745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Freud AG, Yu J, Caligiuri MA. Human natural killer cell development in secondary lymphoid tissues. Semin Immunol. 2014;26:132–7. doi: 10.1016/j.smim.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Valenzuela F, et al. Effects of tofacitinib on lymphocyte sub-populations, CMV and EBV viral load in patients with plaque psoriasis. BMC Dermatol. 2015;15:8. doi: 10.1186/s12895-015-0025-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.van Vollenhoven RF, et al. Relationship Between NK Cell Count and Important Safety Events in Rheumatoid Arthritis Patients Treated with Tofacitinib. Annals of the Rheumatic Diseases. 2015 [Google Scholar]
- 89.Winthrop KL, et al. The effect of tofacitinib on pneumococcal and influenza vaccine responses in rheumatoid arthritis. Ann Rheum Dis. 2015 doi: 10.1136/annrheumdis-2014-207191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Vincenti F, et al. Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant. 2012;12:2446–56. doi: 10.1111/j.1600-6143.2012.04127.x. [DOI] [PubMed] [Google Scholar]
- 91.Migita K, et al. Effects of Janus kinase inhibitor tofacitinib on circulating serum amyloid A and interleukin-6 during treatment for rheumatoid arthritis. Clin Exp Immunol. 2014;175:208–14. doi: 10.1111/cei.12234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Scheller J, Garbers C, Rose-John S. Interleukin-6: from basic biology to selective blockade of pro-inflammatory activities. Semin Immunol. 2014;26:2–12. doi: 10.1016/j.smim.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 93.Souto A, et al. Lipid profile changes in patients with chronic inflammatory arthritis treated with biologic agents and tofacitinib in randomized clinical trials: a systematic review and meta-analysis. Arthritis Rheumatol. 2015;67:117–27. doi: 10.1002/art.38894. [DOI] [PubMed] [Google Scholar]
- 94.Kume K. Tofacitinib Improves Arterial Stiffness Despite up-Regulating Serum Cholesterol with Chronic Cardiovascular Disease in Methotrexate-Resistant Active Rheumatoid Arthritis Patients. a Cohort Study. Arthritis and Rheumatism. 2014;66:S209. [Google Scholar]
- 95.Charles-Schoeman C, et al. Potential mechanisms leading to the abnormal lipid profile in patients with rheumatoid arthritis versus healthy volunteers and reversal by tofacitinib. Arthritis Rheumatol. 2015;67:616–25. doi: 10.1002/art.38974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Moisan A, et al. White-to-brown metabolic conversion of human adipocytes by JAK inhibition. Nat Cell Biol. 2015;17:57–67. doi: 10.1038/ncb3075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases–elimination, equilibrium and escape. Curr Opin Immunol. 2014;27:16–25. doi: 10.1016/j.coi.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Administration, F.a.D. in FDA Press Release (http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm251572.htm, 2011)
- 99.Genentech. in Press Release (http://www.gene.com/media/press-releases/14387/2013-04-30/fda-approves-actemra-for-children-living, 2013)
- 100.Salvarani C, et al. Tocilizumab: a novel therapy for patients with large-vessel vasculitis. Rheumatology (Oxford) 2012;51:151–6. doi: 10.1093/rheumatology/ker296. [DOI] [PubMed] [Google Scholar]
- 101.Unizony S, et al. Tocilizumab for the treatment of large-vessel vasculitis (giant cell arteritis, Takayasu arteritis) and polymyalgia rheumatica. Arthritis Care Res (Hoboken) 2012;64:1720–9. doi: 10.1002/acr.21750. [DOI] [PubMed] [Google Scholar]
- 102.Okuda Y, et al. Comparison of the clinical utility of tocilizumab and anti-TNF therapy in AA amyloidosis complicating rheumatic diseases. Mod Rheumatol. 2014;24:137–43. doi: 10.3109/14397595.2013.854048. [DOI] [PubMed] [Google Scholar]
- 103.Sieper J, Porter-Brown B, Thompson L, Harari O, Dougados M. Assessment of short-term symptomatic efficacy of tocilizumab in ankylosing spondylitis: results of randomised, placebo-controlled trials. Ann Rheum Dis. 2014;73:95–100. doi: 10.1136/annrheumdis-2013-203559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Elhai M, et al. Outcomes of patients with systemic sclerosis-associated polyarthritis and myopathy treated with tocilizumab or abatacept: a EUSTAR observational study. Ann Rheum Dis. 2013;72:1217–20. doi: 10.1136/annrheumdis-2012-202657. [DOI] [PubMed] [Google Scholar]
- 105.Narazaki M, et al. Therapeutic effect of tocilizumab on two patients with polymyositis. Rheumatology (Oxford) 2011;50:1344–6. doi: 10.1093/rheumatology/ker152. [DOI] [PubMed] [Google Scholar]
- 106.Jabbari A, et al. Reversal of Alopecia Areata Following Treatment With the JAK1/2 Inhibitor Baricitinib. EBioMedicine. 2015;2:351–355. doi: 10.1016/j.ebiom.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Craiglow BG, King BA. Killing two birds with one stone: oral tofacitinib reverses alopecia universalis in a patient with plaque psoriasis. J Invest Dermatol. 2014;134:2988–90. doi: 10.1038/jid.2014.260. [DOI] [PubMed] [Google Scholar]
- 108.Pieri L, Guglielmelli P, Vannucchi AM. Ruxolitinib-induced reversal of alopecia universalis in a patient with essential thrombocythemia. Am J Hematol. 2015;90:82–3. doi: 10.1002/ajh.23871. [DOI] [PubMed] [Google Scholar]
- 109.Xing L, et al. Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat Med. 2014;20:1043–9. doi: 10.1038/nm.3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Craiglow BG, King BA, Tofacitinib Citrate. for the Treatment of Vitiligo: A Pathogenesis-Directed Therapy. JAMA Dermatol. 2015 doi: 10.1001/jamadermatol.2015.1520. [DOI] [PubMed] [Google Scholar]
- 111.Harel S, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Science Advances. 2015;1:e1500973–e1500973. doi: 10.1126/sciadv.1500973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Bennett L, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003;197:711–23. doi: 10.1084/jem.20021553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Banchereau J, Pascual V. Type I interferon in systemic lupus erythematosus and other autoimmune diseases. Immunity. 2006;25:383–92. doi: 10.1016/j.immuni.2006.08.010. [DOI] [PubMed] [Google Scholar]
- 114.Hornung T, et al. Remission of recalcitrant dermatomyositis treated with ruxolitinib. N Engl J Med. 2014;371:2537–8. doi: 10.1056/NEJMc1412997. [DOI] [PubMed] [Google Scholar]
- 115.Liu Y, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371:507–18. doi: 10.1056/NEJMoa1312625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Montealegre Sanchez GA, et al. Lipodystrophy and Elevated Temperatures (CANDLE): Clinical Characterization and Initial Response To Janus Kinase Inhibition With Baricitinib. Annals of the Rheumatic Diseases. 2013;65:S758–S759. [Google Scholar]
- 117.Liew SH, et al. Tofacitinib (CP-690,550), a Janus kinase inhibitor for dry eye disease: results from a phase 1/2 trial. Ophthalmology. 2012;119:1328–35. doi: 10.1016/j.ophtha.2012.01.028. [DOI] [PubMed] [Google Scholar]
- 118.Vanhoutte FP, et al. OP0263 Efficacy and safety of GLPG0634, a selective JAK1 inhibitor, after short-term treatment of rheumatoid arthritis; results of a phase IIA trial. Annals of the Rheumatic Diseases. 2014;71:145–145. [Google Scholar]
- 119.Fleischmann RM, et al. A randomized, double-blind, placebo-controlled, twelve-week, dose-ranging study of decernotinib, an oral selective JAK-3 inhibitor, as monotherapy in patients with active rheumatoid arthritis. Arthritis Rheumatol. 2015;67:334–43. doi: 10.1002/art.38949. [DOI] [PubMed] [Google Scholar]
- 120.Genovese MC, van Vollenhoven RF, Pacheco-Tena C, Zhang Y, Kinnman N. VX-509 (Decernotinib), an Oral Selective Janus Kinase 3 Inhibitor, in Combination With Methotrexate in Patients With Rheumatoid Arthritis. Arthritis Rheumatol. 2015 doi: 10.1002/art.39473. [DOI] [PubMed] [Google Scholar]
- 121.London N, et al. Covalent docking of large libraries for the discovery of chemical probes. Nat Chem Biol. 2014;10:1066–72. doi: 10.1038/nchembio.1666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Koo MY, et al. Selective inhibition of the function of tyrosine-phosphorylated STAT3 with a phosphorylation site-specific intrabody. Proc Natl Acad Sci U S A. 2014;111:6269–74. doi: 10.1073/pnas.1316815111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Santoni M, et al. Investigational therapies targeting signal transducer and activator of transcription 3 for the treatment of cancer. Expert Opin Investig Drugs. 2015;24:809–24. doi: 10.1517/13543784.2015.1020370. [DOI] [PubMed] [Google Scholar]

